Metabolic Reprogramming of Pancreatic Ductal Adenocarcinoma Cells in Response to Chronic Low Ph Stress

Total Page:16

File Type:pdf, Size:1020Kb

Metabolic Reprogramming of Pancreatic Ductal Adenocarcinoma Cells in Response to Chronic Low Ph Stress University of Nebraska Medical Center DigitalCommons@UNMC Theses & Dissertations Graduate Studies Fall 12-15-2017 Metabolic Reprogramming of Pancreatic Ductal Adenocarcinoma Cells in Response to Chronic Low pH Stress Jaime Abrego University of Nebraska Medical Center Follow this and additional works at: https://digitalcommons.unmc.edu/etd Part of the Biochemistry Commons, Cancer Biology Commons, Molecular Biology Commons, and the Molecular, Genetic, and Biochemical Nutrition Commons Recommended Citation Abrego, Jaime, "Metabolic Reprogramming of Pancreatic Ductal Adenocarcinoma Cells in Response to Chronic Low pH Stress" (2017). Theses & Dissertations. 249. https://digitalcommons.unmc.edu/etd/249 This Thesis is brought to you for free and open access by the Graduate Studies at DigitalCommons@UNMC. It has been accepted for inclusion in Theses & Dissertations by an authorized administrator of DigitalCommons@UNMC. For more information, please contact [email protected]. Metabolic Reprogramming of Pancreatic Ductal Adenocarcinoma Cells in Response to Chronic Low pH Stress by Jaime Abrego A Dissertation Presented to the Faculty Of the University Of Nebraska Graduate College in Partial Fulfillment of the Requirements for the Degree of Doctor Of Philosophy Cancer Research Graduate Program Under the Supervision of Professor Pankaj K. Singh, Ph.D. University of Nebraska Medical Center Omaha, NE November, 2017 Supervisory Committee: Michael A. (Tony) Hollingsworth, Ph.D. Keith R. Johnson, Ph.D. Amarnath Natarajan, Ph.D. ii iii Acknowledgements First of all, I would like to thank my supervisor and mentor Dr. Pankaj K. Singh for his guidance, support, and motivation during the course of my PhD. I greatly appreciate his leadership, patience, and mentorship that have allowed me to become a better scientist over the years I worked under his tutelage. I am very thankful for the freedom and encouragement to pursue research projects fit to my scientific inquiry. For these reasons, as well as, all the learning experiences, I am indebted to him for giving me the opportunity to work in his lab and begin my journey to investigate the role of metabolism in cancer. I would also like to thank my supervisory committee members Dr. Michael A. (Tony) Hollingsworth, Dr. Keith R. Johnson, and Dr. Amarnath Natarajan for their excellent supervision, suggestions, and constructive criticism throughout my graduate career. Without their guidance, I would not have been able to identify and overcome many significant obstacles throughout my studies. I would also like to thank the current and former members of the Singh lab for their help in learning/optimizing laboratory techniques, as well as, for their moral and intellectual support throughout the years. I would like to especially acknowledge Dr. Venugopal Gunda for guiding my learning of metabolomics studies. The students and faculty of the Eppley Institute have made my personal endeavor through graduate school an enlightening and entertaining time. Moreover, I learned more than I could have ever imagined from my interactions with fellow students and faculty. Their kindness and generosity will always be cherished. Finally, I would like to thank friends and family for their unconditional love and support throughout my life and academic career. They have kept me focused and upbeat throughout the good times and the bad. Without their support and understanding, iv I would have never been able to complete my journey in graduate school. I would like to dedicate this thesis to my parents, Jaime & Irma Abrego, who brought my sisters and me to the United States from Mexico 13 years ago with the goal of giving us the opportunity for a brighter future. My dad works in a warehouse for over 50 hours per week and my mom works at a factory 4 days out of the week—and I cannot thank them enough for the work they have put to raise our family. They have raised us to be hard working, honest, and despite our economic limitations they’ve worked to give us the opportunity to attain higher education. It is because of them that I was able to conduct the research described in this thesis and for this reason this thesis belongs to them as well. v Metabolic Reprogramming of Pancreatic Ductal Adenocarcinoma Cells In Response To Chronic Low pH Stress Jaime Abrego, Ph.D. University of Nebraska Medical Center, 2017 Supervisor: Pankaj K. Singh, Ph.D. Pancreatic ductal adenocarcinoma (PDAC) is among the most lethal of all cancers with a 5-year survival rate of only 8.2%. This is because PDAC is diagnosed in its advanced stages and is characterized by radio and chemotherapy resistance. Aggressiveness of PDAC tumors is attributed to its high metabolic phenotype, which is characterized by increased glycolysis rate and lactate secretion, while oxidative metabolism is reduced. These metabolic features are required to fulfill the biosynthetic demands of proliferating PDAC cells. However, this increase in metabolic activity results in acidification of the extracellular space because the dense fibrotic stroma of PDAC tumors limits venting of protons into the vasculature thereby creating a chronic low pH microenvironment. Little is known regarding the physiology and metabolism of cancer cells enduring chronic low pH exposure. To demonstrate effects of low pH, PDAC cells were cultured in low pH 6.9~7.0 to establish chronic low pH as it occurs in tumors. These cells were compared to cells in physiological pH of 7.4, which is also the pH of cell culture, in order to evaluate physiological differences between these pH values. In these experiments, it was observed that cells in low pH have reduced clonogenic capacity and undergo a metabolic shift to oxidative metabolism that is supported by an increase in glutamine uptake. These observations exhibit a robust contrast to PDAC cells in control pH conditions that are highly glycolytic. Furthermore, in low pH there is increased vi transcription of the GOT1 enzyme, which mediates metabolic flux through the non- canonical glutamine metabolic pathway that allows synthesis of other metabolic substrates from glutamine. Upon shRNA-mediated depletion of GOT1, survival of PDAC cells in low pH was significantly impaired due to increase in ROS to cytotoxic levels. However, supplementing transfected clones with GOT1 metabolic product, oxaloacetate, resulted in growth rescue and reduction in ROS levels. Thus, in chronic low pH stress PDAC cells up-regulate non-canonical glutamine metabolism through increased transcription of GOT1, which allows PDAC cells to generate energy and metabolic co- factors to suppress cytotoxic ROS levels. Low pH is a universal feature of the PDAC tumor microenvironment and further dissection of metabolic adaptations to microenvironment conditions will result in more effective therapy for PDAC. vii Table of Contents Acknowledgements………………………….......................................................................iii Abstract……………............................................................................................................v Table of Contents……………...........................................................................................vii List of Figures……............................................................................................................ix List of Abbreviations……..................................................................................................xi Chapter 1: Introduction: The Evolution Of PDAC And Its Acidic Tumor Microenvironment…………………………….......................................................................1 Incidence of Pancreatic Ductal Adenocarcinoma..............................................................2 Development of Pancreatic Ductal Adenocarcinoma........................................................2 Features of Pancreatic Ductal Adenocarcinoma.............................................................14 Cellular Metabolism………………....................................................................................21 Cancer Cell Metabolism…………....................................................................................28 Tumor Microenvironment Acidification….........................................................................39 Hypothesis.……………....................................................................................................44 Figures Chapter 1……………..........................................................................................46 Tables Chapter 1……………...........................................................................................61 viii Chapter 2: Results: Got1-Mediated Anaplerotic Glutamine Metabolism Regulates Chronic Low pH Stress in PDAC Cells……………………………...................................64 Introduction………….......................................................................................................65 Materials & Methods…....................................................................................................67 Results…………..............................................................................................................76 Discussion………….........................................................................................................82 Chapter 2 Figures…........................................................................................................85 Chapter 3: Discussion: Conclusions & Future Directions.............................................103 Summary of Work…......................................................................................................104
Recommended publications
  • COX4I2 Antibody (Aa31-80) Rabbit Polyclonal Antibody Catalog # ALS15898
    10320 Camino Santa Fe, Suite G San Diego, CA 92121 Tel: 858.875.1900 Fax: 858.622.0609 COX4I2 Antibody (aa31-80) Rabbit Polyclonal Antibody Catalog # ALS15898 Specification COX4I2 Antibody (aa31-80) - Product Information Application IHC, IF, WB Primary Accession Q96KJ9 Reactivity Human, Mouse, Rat Host Rabbit Clonality Polyclonal Calculated MW 20kDa KDa COX4I2 Antibody (aa31-80) - Additional Information Gene ID 84701 Anti-COX4I2 antibody IHC staining of human kidney. Other Names Cytochrome c oxidase subunit 4 isoform 2, mitochondrial, Cytochrome c oxidase subunit IV isoform 2, COX IV-2, COX4I2, COX4L2 Target/Specificity COX42 Antibody detects endogenous levels Immunofluorescence of COS7 cells, using of total COX42 protein. COX42 Antibody. Reconstitution & Storage Store at -20°C for up to one year. Precautions COX4I2 Antibody (aa31-80) is for research use only and not for use in diagnostic or therapeutic procedures. COX4I2 Antibody (aa31-80) - Protein Information Western blot of extracts from K562 cells, Name COX4I2 treated with insulin 0.01U/ml 15', using COX42 Antibody. Synonyms COX4L2 Function COX4I2 Antibody (aa31-80) - Background Component of the cytochrome c oxidase, the last enzyme in the mitochondrial This protein is one of the nuclear-coded electron transport chain which drives polypeptide chains of cytochrome c oxidase, oxidative phosphorylation. The respiratory the terminal oxidase in mitochondrial electron chain contains 3 multisubunit complexes transport. Page 1/2 10320 Camino Santa Fe, Suite G San Diego, CA 92121 Tel: 858.875.1900 Fax: 858.622.0609 succinate dehydrogenase (complex II, CII), COX4I2 Antibody (aa31-80) - References ubiquinol- cytochrome c oxidoreductase (cytochrome b-c1 complex, complex III, CIII) Huettemann M.,et al.Gene and cytochrome c oxidase (complex IV, 267:111-123(2001).
    [Show full text]
  • Mutation in the COX4I1 Gene Is Associated with Short Stature, Poor Weight Gain and Increased Chromosomal Breaks, Simulating Fanconi Anemia
    European Journal of Human Genetics (2017) 25, 1142–1146 & 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved 1018-4813/17 www.nature.com/ejhg ARTICLE Mutation in the COX4I1 gene is associated with short stature, poor weight gain and increased chromosomal breaks, simulating Fanconi anemia Bassam Abu-Libdeh*,1,4, Liza Douiev2,3,4, Sarah Amro1, Maher Shahrour1, Asaf Ta-Shma2, Chaya Miller2,3, Orly Elpeleg2 and Ann Saada*,2,3 We describe a novel autosomal recessive form of mitochondrial disease in a child with short stature, poor weight gain, and mild dysmorphic features with highly suspected Fanconi anemia due to a mutation in COX4I1 gene. Whole Exome Sequencing was performed then followed by Sanger confirmation, identified a K101N mutation in COX4I1, segregating with the disease. This nuclear gene encodes the common isoform of cytochrome c oxidase (COX) subunit 4 (COX 4-1), an integral regulatory part of COX (respiratory chain complex IV) the terminal electron acceptor of the mitochondrial respiratory chain. The patient’s fibroblasts disclosed decreased COX activity, impaired ATP production, elevated ROS production, decreased expression of COX4I1 mRNA and undetectable (COX4) protein. COX activity and ATP production were restored by lentiviral transfection with the wild-type gene. Our results demonstrate the first human mutation in the COX4I1 gene linked to diseases and confirm its role in the pathogenesis. Thus COX4I1 mutations should be considered in any patient with features suggestive of this diagnosis. European Journal of Human Genetics (2017) 25, 1142–1146; doi:10.1038/ejhg.2017.112; published online 2 August 2017 INTRODUCTION normal vaginal delivery with birth weight of 2.8 kg after an uneventful Mitochondrial cytochrome c oxidase (COX, complex IV) is the terminal pregnancy.
    [Show full text]
  • Loss of COX4I1 Leads to Combined Respiratory Chain Deficiency And
    cells Article Loss of COX4I1 Leads to Combined Respiratory Chain Deficiency and Impaired Mitochondrial Protein Synthesis Kristýna Cunˇ átová 1,2, David Pajuelo Reguera 1 , Marek Vrbacký 1, Erika Fernández-Vizarra 3 , Shujing Ding 3, Ian M. Fearnley 3, Massimo Zeviani 3 , Josef Houštˇek 1 , Tomáš Mráˇcek 1,* and Petr Pecina 1,* 1 Laboratory of Bioenergetics, Institute of Physiology, Czech Academy of Sciences, 142 00 Prague, Czech Republic; [email protected] (K.C.);ˇ [email protected] (D.P.R.); [email protected] (M.V.); [email protected] (J.H.) 2 Department of Cell Biology, Faculty of Science, Charles University, 128 00 Prague, Czech Republic 3 MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK; [email protected] (E.F.-V.); [email protected] (S.D.); [email protected] (I.M.F.); [email protected] (M.Z.) * Correspondence: [email protected] (T.M.); [email protected] (P.P.) Abstract: The oxidative phosphorylation (OXPHOS) system localized in the inner mitochondrial membrane secures production of the majority of ATP in mammalian organisms. Individual OXPHOS complexes form supramolecular assemblies termed supercomplexes. The complexes are linked not only by their function but also by interdependency of individual complex biogenesis or maintenance. For instance, cytochrome c oxidase (cIV) or cytochrome bc1 complex (cIII) deficiencies affect the level of fully assembled NADH dehydrogenase (cI) in monomeric as well as supercomplex forms. It was hypothesized that cI is affected at the level of enzyme assembly as well as at the level of cI stability and maintenance.
    [Show full text]
  • Hypoxia Inducible Factors Modulate Mitochondrial Oxygen Consumption and Transcriptional Regulation of Nuclear-Encoded Electron T
    Article pubs.acs.org/biochemistry Hypoxia Inducible Factors Modulate Mitochondrial Oxygen Consumption and Transcriptional Regulation of Nuclear-Encoded Electron Transport Chain Genes † § † ‡ ∥ † † ‡ † Hye Jin Hwang, , Scott G. Lynn, , , Ajith Vengellur, Yogesh Saini, , Elizabeth A. Grier, † § † § ‡ Shelagh M. Ferguson-Miller, , and John J. LaPres*, , , † ‡ § Department of Biochemistry and Molecular Biology, Center for Integrative Toxicology, and Center for Mitochondrial Science and Medicine, Michigan State University, East Lansing, Michigan 48824-1319, United States *S Supporting Information ABSTRACT: Hypoxia inducible factor-1 (HIF1) is a stress-responsive nuclear transcription factor that is activated with a decrease in oxygen availability. HIF1 regulates the expression of genes involved in a cell’s adaptation to hypoxic stress, including those with mitochondrial specific function. To gain a more comprehensive understanding of the role of HIF1 in mitochondrial homeostasis, we studied the link between hypoxia, HIF1 transactivation, and electron transport chain (ETC) function. We established immortalized mouse embryonic fibroblasts (MEFs) for HIF1α wild-type (WT) and null cells and tested whether HIF1α regulates mitochondrial respiration by modulating gene expressions of nuclear-encoded ETC components. High- throughput quantitative real-time polymerase chain reaction was performed to screen nuclear- encoded mitochondrial genes related to the ETC to identify those whose regulation was HIF1α- dependent. Our data suggest that HIF1α regulates transcription of cytochrome c oxidase (CcO) heart/muscle isoform 7a1 (Cox7a1) under hypoxia, where it is induced 1.5−2.5-fold, whereas Cox4i2 hypoxic induction was HIF1α-independent. We propose that adaptation to hypoxic stress of CcO as the main cellular oxygen consumer is mediated by induction of hypoxia-sensitive tissue-specific isoforms.
    [Show full text]
  • COX4I2 Polyclonal Antibody
    For Research Use Only COX4I2 Polyclonal antibody Catalog Number:11463-1-AP Featured Product 12 Publications www.ptgcn.com Catalog Number: GenBank Accession Number: Recommended Dilutions: Basic Information 11463-1-AP BC057779 WB 1:500-1:3000 Size: GeneID (NCBI): IP 0.5-4.0 ug for IP and 1:500-1:2000 850 μg/ml 84701 for WB IHC 1:20-1:200 Source: Full Name: IF 1:20-1:200 Rabbit cytochrome c oxidase subunit IV Isotype: isoform 2 (lung) IgG Calculated MW: Purification Method: 171 aa, 20 kDa Antigen affinity purification Observed MW: Immunogen Catalog Number: 17-18 kDa AG1987 Applications Tested Applications: Positive Controls: IF, IHC, IP, WB,ELISA WB : HepG2 cells; human brain tissue, MCF-7 cells, Cited Applications: human heart tissue, human lung tissue, A375 cells IF, IHC, WB IP : HepG2 cells; Species Specificity: IHC : human ovary tumor tissue; human, mouse, rat Cited Species: IF : Hela cells; human, mouse, rat Note-IHC: suggested antigen retrieval with TE buffer pH 9.0; (*) Alternatively, antigen retrieval may be performed with citrate buffer pH 6.0 COX4I2, also named as COX4L2 and COXIV-2, belongs to the cytochrome c oxidase IV family. It is one of the Background Information nuclear-coded polypeptide chains of cytochrome c oxidase, the terminal oxidase in mitochondrial electron transport. COXIV(cytochrome c oxidase IV) has two isoforms (isoform 1 and 2). Isoform 1(COX4I1) is ubiquitously expressed and isoform 2 is highly expressed in lung tissues. This antibody was generated against full length COX4I2 protein. Notable Publications Author Pubmed ID Journal Application Siddhesh Aras 27913209 Biochim Biophys Acta WB Guy Keller 30423473 Neurobiol Dis IF Theodoros Karampelas 24661240 Bioconjug Chem WB Storage: Storage Store at -20ºC.
    [Show full text]
  • Blueprint Genetics COX4I2 Single Gene Test
    COX4I2 single gene test Test code: S02924 Phenotype information Exocrine pancreatic insufficiency, dyserythropoietic anemia, and calvarial hyperostosis Alternative gene names COX4-2, COX4B, COXIV-2, dJ857M17.2 Test Strengths The strengths of this test include: CAP accredited laboratory CLIA-certified personnel performing clinical testing in a CLIA-certified laboratory Powerful sequencing technologies, advanced target enrichment methods and precision bioinformatics pipelines ensure superior analytical performance Careful construction of clinically effective and scientifically justified gene panels Our Nucleus online portal providing transparent and easy access to quality and performance data at the patient level Our publicly available analytic validation demonstrating complete details of test performance ~2,000 non-coding disease causing variants in our clinical grade NGS assay for panels (please see ‘Non-coding disease causing variants covered by this test’) Our rigorous variant classification scheme Our systematic clinical interpretation workflow using proprietary software enabling accurate and traceable processing of NGS data Our comprehensive clinical statements Test Limitations This test does not detect the following: Complex inversions Gene conversions Balanced translocations Mitochondrial DNA variants Repeat expansion disorders unless specifically mentioned Non-coding variants deeper than ±20 base pairs from exon-intron boundary unless otherwise indicated (please see above non-coding variants covered by the test). This test may not reliably detect the following: Low level mosaicism (variant with a minor allele fraction of 14.6% is detected with 90% probability) Stretches of mononucleotide repeats Indels larger than 50bp Single exon deletions or duplications Variants within pseudogene regions/duplicated segments The sensitivity of this test may be reduced if DNA is extracted by a laboratory other than Blueprint Genetics.
    [Show full text]
  • Upregulation of COX4-2 Via HIF-1 in Mitochondrial COX4-1 Deficiency
    cells Article Upregulation of COX4-2 via HIF-1α in Mitochondrial COX4-1 Deficiency Liza Douiev 1,*, Chaya Miller 1, Shmuel Ruppo 2 , Hadar Benyamini 2, Bassam Abu-Libdeh 3 and Ann Saada 1,4,* 1 Department of Genetics, Hadassah Medical Center, Jerusalem 9112001, Israel; [email protected] 2 Info-CORE, I-CORE Bioinformatics Unit of the Hebrew, University of Jerusalem and Hadassah Medical Center, Jerusalem 91120011, Israel; [email protected] (S.R.); [email protected] (H.B.) 3 Department of Pediatrics and Genetics, Makassed Hospital and Al-Quds University, East Jerusalem 91220, Palestine; [email protected] 4 Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112001, Israel * Correspondence: [email protected] (L.D.); [email protected] or [email protected] (A.S.) Abstract: Cytochrome-c-oxidase (COX) subunit 4 (COX4) plays important roles in the function, assembly and regulation of COX (mitochondrial respiratory complex 4), the terminal electron acceptor of the oxidative phosphorylation (OXPHOS) system. The principal COX4 isoform, COX4-1, is expressed in all tissues, whereas COX4-2 is mainly expressed in the lungs, or under hypoxia and other stress conditions. We have previously described a patient with a COX4-1 defect with a relatively mild presentation compared to other primary COX deficiencies, and hypothesized that this could be the result of a compensatory upregulation of COX4-2. To this end, COX4-1 was downregulated by shRNAs in human foreskin fibroblasts (HFF) and compared to the patient’s cells. COX4-1, COX4-2 and HIF-1α were detected by immunocytochemistry.
    [Show full text]
  • Goat Anti-COX4I2 & COX4I1 Antibody
    10320 Camino Santa Fe, Suite G San Diego, CA 92121 Tel: 858.875.1900 Fax: 858.622.0609 Goat Anti-COX4I2 & COX4I1 Antibody Peptide-affinity purified goat antibody Catalog # AF2173a Specification Goat Anti-COX4I2 & COX4I1 Antibody - Product Information Application WB Primary Accession P13073 Other Accession NP_115998, 84701, 1327, 84682 (mouse), 84683 (rat) Reactivity Human Predicted Mouse, Rat, Dog Host Goat Clonality Polyclonal Concentration 0.5 mg/ml AF2173a (1 µg/ml) staining of Human Isotype IgG Skeletal Muscle lysate (35 µg protein in RIPA Calculated MW 19577 buffer). Primary incubation was 1 hour. Detected by chemiluminescence. Goat Anti-COX4I2 & COX4I1 Antibody - Additional Information Goat Anti-COX4I2 & COX4I1 Antibody - Background Gene ID 1327 Cytochrome c oxidase (COX), the terminal enzyme of the mitochondrial respiratory chain, Other Names catalyzes the electron transfer from reduced Cytochrome c oxidase subunit 4 isoform 1, mitochondrial, Cytochrome c oxidase cytochrome c to oxygen. It is a heteromeric polypeptide IV, Cytochrome c oxidase complex consisting of 3 catalytic subunits subunit IV isoform 1, COX IV-1, COX4I1, encoded by mitochondrial genes and multiple COX4 structural subunits encoded by nuclear genes. The mitochondrially-encoded subunits function Format in electron transfer, and the nuclear-encoded 0.5 mg IgG/ml in Tris saline (20mM Tris subunits may be involved in the regulation and pH7.3, 150mM NaCl), 0.02% sodium azide, assembly of the complex. This nuclear gene with 0.5% bovine serum albumin encodes isoform 2 of subunit IV. Isoform 1 of subunit IV is encoded by a different gene, Storage however, the two genes show a similar Maintain refrigerated at 2-8°C for up to 6 structural organization.
    [Show full text]
  • Genome-Wide Identification of Mrnas Associated with the Protein SMN Whose Depletion Decreases Their Axonal Localization
    Downloaded from rnajournal.cshlp.org on October 2, 2021 - Published by Cold Spring Harbor Laboratory Press Genome-wide identification of mRNAs associated with the protein SMN whose depletion decreases their axonal localization FLORENCE RAGE,1,2,3 NAWAL BOULISFANE,1,2,3 KHALIL RIHAN,1,2,3 HENRY NEEL,1,2,3,4 THIERRY GOSTAN,1,2,3 EDOUARD BERTRAND,1,2,3 RÉMY BORDONNÉ,1,2,3 and JOHANN SORET1,2,3,5 1Institut de Génétique Moléculaire de Montpellier UMR 5535, 34293 Montpellier Cedex 5, France 2Université Montpellier 2, 34095 Montpellier Cedex 5, France 3Université Montpellier 1, 34967 Montpellier Cedex 2, France ABSTRACT Spinal muscular atrophy is a neuromuscular disease resulting from mutations in the SMN1 gene, which encodes the survival motor neuron (SMN) protein. SMN is part of a large complex that is essential for the biogenesis of spliceosomal small nuclear RNPs. SMN also colocalizes with mRNAs in granules that are actively transported in neuronal processes, supporting the hypothesis that SMN is involved in axonal trafficking of mRNPs. Here, we have performed a genome-wide analysis of RNAs present in complexes containing the SMN protein and identified more than 200 mRNAs associated with SMN in differentiated NSC-34 motor neuron-like cells. Remarkably, ∼30% are described to localize in axons of different neuron types. In situ hybridization and immuno-fluorescence experiments performed on several candidates indicate that these mRNAs colocalize with the SMN protein in neurites and axons of differentiated NSC-34 cells. Moreover, they localize in cell processes in an SMN-dependent manner. Thus, low SMN levels might result in localization deficiencies of mRNAs required for axonogenesis.
    [Show full text]
  • Role of Cytochrome C Oxidase Nuclear-Encoded Subunits in Health and Disease
    Physiol. Res. 69: 947-965, 2020 https://doi.org/10.33549/physiolres.934446 REVIEW Role of Cytochrome c Oxidase Nuclear-Encoded Subunits in Health and Disease Kristýna ČUNÁTOVÁ1, David PAJUELO REGUERA1, Josef HOUŠTĚK1, Tomáš MRÁČEK1, Petr PECINA1 1Department of Bioenergetics, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic Received February 2, 2020 Accepted September 13, 2020 Epub Ahead of Print November 2, 2020 Summary [email protected] and Tomáš Mráček, Department of Cytochrome c oxidase (COX), the terminal enzyme of Bioenergetics, Institute of Physiology CAS, Vídeňská 1083, 142 mitochondrial electron transport chain, couples electron transport 20 Prague 4, Czech Republic. E-mail: [email protected] to oxygen with generation of proton gradient indispensable for the production of vast majority of ATP molecules in mammalian Cytochrome c oxidase cells. The review summarizes current knowledge of COX structure and function of nuclear-encoded COX subunits, which may Energy demands of mammalian cells are mainly modulate enzyme activity according to various conditions. covered by ATP synthesis carried out by oxidative Moreover, some nuclear-encoded subunits possess tissue-specific phosphorylation apparatus (OXPHOS) located in the and development-specific isoforms, possibly enabling fine-tuning central bioenergetic organelle, mitochondria. OXPHOS is of COX function in individual tissues. The importance of nuclear- composed of five multi-subunit complexes embedded in encoded subunits is emphasized by recently discovered the inner mitochondrial membrane (IMM). Electron pathogenic mutations in patients with severe mitopathies. In transport from reduced substrates of complexes I and II to addition, proteins substoichiometrically associated with COX were cytochrome c oxidase (COX, complex IV, CIV) is found to contribute to COX activity regulation and stabilization of achieved by increasing redox potential of individual the respiratory supercomplexes.
    [Show full text]
  • Discovery of Cancer Common and Specific Driver Gene Sets
    1 Discovery of cancer common and specific driver gene sets Junhua Zhang and Shihua Zhang Abstract—Cancer is known as a disease mainly caused by gene alterations. Discovery of mutated driver pathways or gene sets is becoming an important step to understand molecular mechanisms of carcinogenesis. However, systematically investigating commonalities and specificities of driver gene sets among multiple cancer types is still a great challenge, but this investigation will undoubtedly benefit deciphering cancers and will be helpful for personalized therapy and precision medicine in cancer treatment. In this study, we propose two optimization models to de novo discover common driver gene sets among multiple cancer types (ComMDP) and specific driver gene sets of one certain or multiple cancer types to other cancers (SpeMDP), respectively. We first apply ComMDP and SpeMDP to simulated data to validate their efficiency. Then, we further apply these methods to 12 cancer types from The Cancer Genome Atlas (TCGA) and obtain several biologically meaningful driver pathways. As examples, we construct a common cancer pathway model for BRCA and OV, infer a complex driver pathway model for BRCA carcinogenesis based on common driver gene sets of BRCA with eight cancer types, and investigate specific driver pathways of the liquid cancer lymphoblastic acute myeloid leukemia (LAML) versus other solid cancer types. In these processes more candidate cancer genes are also found. Index Terms—Bioinformatics, cancer genomics, pan-cancer study, driver pathway, mutual exclusivity ✦ 1 INTRODUCTION metastasis [14], [15]. A main concern is that the human ANCER is a complex and heterogeneous disease with protein interaction network and biological pathways are C diverse genetic and environmental factors involved in far from being complete.
    [Show full text]
  • Cox4i2 Triggers an Increase in Reactive Oxygen Species, Leading to Ferroptosis and Apoptosis in HHV7 Infected Schwann Cells
    fmolb-08-660072 April 30, 2021 Time: 20:29 # 1 ORIGINAL RESEARCH published: 07 May 2021 doi: 10.3389/fmolb.2021.660072 Cox4i2 Triggers an Increase in Reactive Oxygen Species, Leading to Ferroptosis and Apoptosis in HHV7 Infected Schwann Cells Bowen Chang1†, Haochen Guan2†, Xueyi Wang1, Zheng Chen1, Wanchun Zhu1, Xiangyu Wei1 and Shiting Li1* 1 Department of Neurosurgery, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China, 2 Department of Nephrology, School of Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China Emerging evidence suggests that reactive oxygen species (ROS) play a significant role in the pathogenesis of peripheral nerve damage. Our previous study indicated that human herpesvirus 7 (HHV7) induces Bell’s palsy. However, the specific mechanism underlying Edited by: the effects of ROS in HHV7 infection-induced facial nerve damage is unknown. In Fernando Antunes, this study, we established a rat FN model by inoculating an HHV7 virus solution. University of Lisbon, Portugal The facial grading score and LuxolFastBlue (LFB) staining were used to assess the Reviewed by: Ana Cipak Gasparovic, success of the model. Using mRNA-sequencing analysis, we found that the expression Rudjer Boskovic Institute, Croatia of Complex IV Subunit 4 Isoform 2 (Cox4i2) increased in infected Schwann cells (SCs). Ana Sofia Fernandes, CBIOS, Universidade Lusófona Cox4i2 was suggested to increase COX activity, thereby promoting ROS production. Research Center for Biosciences The changes in the endogenous oxidant and antioxidant system were assessed, and and Health Technologies, Portugal the results showed that oxidative stress increased after HHV7 infection in vivo and *Correspondence: in vitro.
    [Show full text]